Aptamer-functionalized lipid nanoparticles targeting ...

4 downloads 0 Views 4MB Size Report
Feb 9, 2015 - ... Fuchu He3, Lingqiang Zhang3, Aiping Lu1,2,4–8,15 & Ge Zhang1,2,4–8,13,15 ...... Wang, Y. & Grainger, D.W. RNA therapeutics targeting ...
T e c h n i c a l R e p o rt s

© 2015 Nature America, Inc. All rights reserved.

Aptamer-functionalized lipid nanoparticles targeting osteoblasts as a novel RNA interference–based bone anabolic strategy Chao Liang1–8,22, Baosheng Guo1,2,4–8,22, Heng Wu1,4–7,22, Ningsheng Shao9, Defang Li1,4–8, Jin Liu1,4–8, Lei Dang1,4–8, Cheng Wang1,10, Hui Li9, Shaohua Li9, Wing Ki Lau1, Yu Cao3, Zhijun Yang1,4–7, Cheng Lu1,2,4–8, Xiaojuan He1,2,4–7, D W T Au11, Xiaohua Pan1, Bao-Ting Zhang12, Changwei Lu1, Hongqi Zhang1, Kinman Yue1, Airong Qian8,13, Peng Shang8,13, Jiake Xu14, Lianbo Xiao1,15, Zhaoxiang Bian1,4–7, Weihong Tan1,7,16–21, Zicai Liang4, Fuchu He3, Lingqiang Zhang3, Aiping Lu1,2,4–8,15 & Ge Zhang1,2,4–8,13,15 Currently, major concerns about the safety and efficacy of RNA interference (RNAi)-based bone anabolic strategies still exist because of the lack of direct osteoblast-specific delivery systems for osteogenic siRNAs. Here we screened the aptamer CH6 by cell-SELEX, specifically targeting both rat and human osteoblasts, and then we developed CH6 aptamer–functionalized lipid nanoparticles (LNPs) encapsulating osteogenic pleckstrin homology domain-containing family O member 1 (Plekho1) siRNA (CH6-LNPs-siRNA). Our results showed that CH6 facilitated in vitro osteoblast-selective uptake of Plekho1 siRNA, mainly via macropinocytosis, and boosted in vivo osteoblastspecific Plekho1 gene silencing, which promoted bone formation, improved bone microarchitecture, increased bone mass and enhanced mechanical properties in both osteopenic and healthy rodents. These results indicate that osteoblast-specific aptamerfunctionalized LNPs could act as a new RNAi-based bone anabolic strategy, advancing the targeted delivery selectivity of osteogenic siRNAs from the tissue level to the cellular level. Metabolic skeletal disorders associated with impaired bone formation (for example, osteoporosis) remain major clinical challenges. RNA

interference (RNAi)-based approaches aimed at promoting osteo­ blastic bone formation may hold therapeutic potential1,2. However, a major bottleneck for translating RNAi-based approaches into clinical application is the lack of osteoblast-specific osteogenic siRNA delivery systems3. Plekho1 (also known as casein kinase-2 interacting protein-1 (CKIP-1)) has been identified as an intracellular negative regulator of bone formation that does not affect bone resorption4. Previously we developed a targeting system involving dioleoyl trimethylammonium propane (DOTAP)-based cationic liposomes attached to six repetitive sequences of aspartate, serine and serine ((AspSerSer) 6), which had good affinity for the physiochemical features of the bone-formation surface when compared to the bone-resorption surface. By using this system, osteogenic Plekho1 siRNA was specifically delivered to the bone-formation surface to promote bone formation5. However, as the system was not specific to osteoblasts at the cellular level, other non-osteoblasts near the bone-formation surface, including endothelial cells and lymphocytes, may also be targeted, which arouses concerns about efficacy and potential toxic side effects5–7. In addition, other potential concerns, including mononuclear phagocyte system (MPS)-induced dose reduction8,9, inefficient nanoparticle extravasation

1Institute

for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China. of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China. 3State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China. 4Academician Chen Xinzi Workroom for Advancing Translational Medicine in Bone & Joint Diseases, Kunshan RNAi Institute, Kunshan Industrial Technology Research Institute, Kunshan, Jiangsu, China. 5Institute of Integrated Bioinfomedicine & Translational Science, Hong Kong Baptist University Shenzhen Research Institute and Continuing Education, Shenzhen, China. 6Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, Hong Kong Baptist University Shenzhen Research Institute and Continuing Education, Shenzhen, China. 7Hong Kong Baptist University Branch of State Key Laboratory of Chemo/Biosensing and Chemometrics of Hunan University, Hong Kong, China. 8Hong Kong Baptist University–Northwestern Polytechnical University Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Shenzhen, China. 9Department of Biochemistry and Molecular Biology, Beijing Institute of Basic Medical Science, Beijing, China. 10Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China. 11Department of Biology and Chemistry, City University of Hong Kong, Hong Kong SAR, China. 12School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China. 13Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Science, Northwestern Polytechnical University, Xi’an, China. 14Molecular Laboratory, School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Australia. 15Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China. 16Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China. 17College of Biology, Hunan University, Changsha, China. 18Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University, Changsha, China. 19Department of Chemistry, University of Florida, Gainesville, Florida, USA. 20Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA. 21Center for Research at Bio/Nano Interface, Shands Cancer Center, University of Florida, Gainesville, Florida, USA. 22These authors contributed equally to this work. Correspondence should be addressed to G.Z. ([email protected]), A.L. ([email protected]) or L.Z. ([email protected]). 2Institute

Received 6 February 2014; accepted 2 December 2014; published online 9 February 2015; doi:10.1038/nm.3791

nature medicine  advance online publication



BRL-3A

Events 0 100 101 102 103 Fluorescence intensity (AU)

350 Events

350

0 100 101 102 103 Fluorescence intensity (AU)

0

Rat PBMCs Library CH2 CH5 CH6

100 101 102 103 Fluorescence intensity (AU)

80

Library CH2

40

CH5

* *

0

350

350

100 101 102 103 Fluorescence intensity (AU)

*

* **

* *

Rat OBs

0

**

*

Concentration (nM)

d

20

* *

10

CH6 15 0 20 0 25 0

0 100 101 102 103 Fluorescence intensity (AU)

120

Events

b 350 Rat OBs

Library 6th pool 10th pool 14th pool

Events 0 100 101 102 103 Fluorescence intensity (AU)

e

c

Rat PBMCs

10 0

0 100 101 102 103 Fluorescence intensity (AU)

350

0

BRL-3A

Events

350

50

Rat OBs

Events

a 350

In vitro analysis of cellular uptake Next, we prepared and characterized CH6 aptamer–functionalized LNPs encapsulating osteogenic Plekho1 siRNA. CH6-LNPs-siRNA had a diameter of 84.0 ± 5.3 nm, encapsulation efficiency above 80%, uniform particle shape, good serum stability, no detectable cytotoxicity and high in vitro osteoblast selectivity (Supplementary Fig. 2; Supplementary Table 2). A random sequence (Rd) served as the negative control aptamer. We labeled Plekho1 siRNA with Cy3 and investigated the effect of the CH6 aptamer versus the Rd aptamer on the cellular uptake of siRNA. The most intense fluorescence signals were detected in osteoblasts incubated with CH6-LNPs-siRNA, and fluorescence signals were rarely observed in osteoblasts treated with no aptamer–conjugated LNPs-siRNA or Rd-LNPs-siRNA (Fig. 2a). Pretreatment of osteoblasts with free CH6 aptamer resulted in a significantly decreased fluorescence signal of CH6-LNPs-siRNA in osteoblasts (P < 0.05; Fig. 2a). We also examined the Plekho1 gene knockdown efficiency of different siRNA formulations in osteoblasts.

Mean fluorescence (AU)

RESULTS Selection of osteoblast-specific aptamers We used rat primary osteoblasts to select aptamers by cell-SELEX from a library composed of 1015 different ssDNA sequences. With increasing rounds of selection we observed progressive enhancement of fluorescence intensity for fluorescein amidite (FAM)-labeled ssDNA pools in target cells (rat primary osteoblasts) by flow cytometry analysis, whereas we observed no obvious change of fluorescence intensity in non-target cells (rat liver cell line BRL-3A and rat PBMCs) (Fig. 1a). After 14 rounds of selection, we sequenced the highly enriched ssDNA pool

and chose 20 representative sequences (from 200 clones), on the basis of their predicted secondary structures, for truncation to remove the non-critical bases17 (Supplementary Table 1). Flow cytometry analysis showed that three aptamer candidates (CH2, CH5 and CH6) had good binding ability to osteoblasts (Fig. 1b) with equilibrium dissociation constants (Kd) in the nanomolar-to-picomolar range (Fig. 1c). To minimize nuclease degradation, CH2, CH5 and CH6 were modified with 2′-O-methyl-nucleotide substitutions18,19 and flow cytometry analysis revealed that they still bound with high affinity to rat osteoblasts but not rat osteoclasts (Fig. 1d). CH2 and CH6 aptamers also bound to human primary osteoblasts but not human osteoclasts or liver cells (THLE-3) (Supplementary Fig. 1). We chose the CH6 aptamer because it had satisfactory secondary structure and shorter nucleotide sequences (Fig. 1e), making it easier to synthesize and conjugate to LNPs-siRNA than CH2.

Events

caused by large particle size (larger than bony sinusoids (80–100 nm))5,10 and detrimental hepatocyte accumulation of siRNA11, should also be considered by drug developers. Here we sought to develop an osteoblast-specific delivery system for osteogenic siRNAs. First we used cell-based systematic evolution of ligands by exponential enrichment (cell-SELEX) to select osteoblastspecific aptamers, which are single-stranded oligonucleotides that use distinct tertiary structures to specifically bind to target cells 12,13. By performing positive selection with osteoblasts and negative selection with hepatocytes and peripheral blood mononuclear cells (PBMCs), we aimed to screen an aptamer that could achieve direct osteoblastspecific delivery of osteogenic siRNAs and minimal hepatocyte and PBMC accumulation of osteogenic siRNAs. Second, using lipid nanoparticles (LNPs), which are